You are on page 1of 12

Virus Evolution, 2021, 7(1): veab032

doi: 10.1093/ve/veab032
Research Article

Downloaded from https://academic.oup.com/ve/article/7/1/veab032/6206434 by Universidade Federal de Viçosa user on 25 March 2022


Interannual dynamics, diversity and evolution of the
virome in Sclerotinia sclerotiorum from a single crop field
Jichun Jia,1,2 Yanping Fu,2 Daohong Jiang,1,2 Fan Mu,1,2 Jiasen Cheng,1,2
Yang Lin,2 Bo Li,1,2 Shin-Yi Lee Marzano,3 and Jiatao Xie1,2,*,†
1
State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, 430070 Hubei
Province, People’s Republic of China, 2Hubei Key Laboratory of Plant Pathology, College of Plant Science and
Technology, Huazhong Agricultural University, Hubei Province, Wuhan 430070, People’s Republic of China
and 3United States Department of Agriculture/Agricultural Research Service, Toledo, OH 43606, USA.
*Corresponding author: E-mail: jiataoxie@mail.hzau.edu.cn

https://orcid.org/0000-0003-1961-0338

Abstract
Mycovirus diversity is generally analyzed from isolates of fungal culture isolates at a single point in time as a snapshot. The
stability of mycovirus composition within the same geographical location over time remains unclear. Not knowing how the
population fluctuates in the field can be a source of unpredictability in the successful application of virocontrol. To better
understand the changes over time, we monitored the interannual dynamics and abundance of mycoviruses infecting
Sclerotinia sclerotiorum at a rapeseed-growing field for three years. We found that the virome in S. sclerotiorum harbors unique
mycovirus compositions each year. In total, sixty-eight mycoviruses were identified, among which twenty-four were
detected in all three successive years. These twenty-four mycoviruses can be classified as the members of the core virome
in this S. sclerotiorum population, which show persistence and relatively high transmissibility under field conditions. Nearly
two-thirds of the mycoviruses have positive-sense, single-stranded RNA genomes and were found consistently across all
three years. Moreover, twenty-eight mycoviruses are newly described, including four novel, multi-segmented narnaviruses,
and four unique bunyaviruses. Overall, the newly discovered mycoviruses in this study belong to as many as twenty
families, into which eight were first identified in S. sclerotiorum, demonstrating evolutionarily diverse viromes. Our findings
not only shed light on the annual variation of mycovirus diversity but also provide important virus evolutionary clues.

Key words: Sclerotinia sclerotiorum; core mycoviruses; interannual dynamics; diversity; evolution.

1. Introduction friendly biological controls, including virocontrol with hypovir-


Many fungi are pathogenic and cause severe disease in a variety ulence-associated mycoviruses, have been considered a viable
of plants and animals, including humans (Sharon and alternative approach (Garcı́a-Pedrajas et al. 2019).
Shlezinger 2013). Phytopathogenic fungi are responsible for ap- Viruses are the most abundant and the most diverse
proximately 70-80% of plant diseases (Oerke 2006). When resis- organisms that exist almost everywhere on Earth (Güemes et al.
tant cultivars are not available, the application of chemical 2016). They infect all types of life forms, from animals and
fungicides is the primary method for controlling fungal diseases plants to microorganisms, including bacteria and fungi (Koonin
in economically important crops. However, environmentally et al. 2006). Metagenomics or metatranscriptomics have

C The Author(s) 2021. Published by Oxford University Press.


V
This is an Open Access article distributed under the terms of the Creative Commons Attribution Non-Commercial License (http://creativecommons.org/
licenses/by-nc/4.0/), which permits non-commercial re-use, distribution, and reproduction in any medium, provided the original work is properly cited.
For commercial re-use, please contact journals.permissions@oup.com

1
2 | Virus Evolution, 2021, Vol. 7, No. 1

revealed a remarkable diversity of viruses sampled from various to obtain 100 isolates of S. sclerotiorum. A total of 300 isolates
cellular organisms in the past few decades (Zhang et al. 2019). were purified for three years. All isolates were cultured on po-
The uncovering of the virosphere diversity has rapidly trans- tato dextrose agar at 20  C and stored at 4  C.
formed our understanding of virus diversity and evolution
(Zhang et al. 2018). A typical virus has a coat protein that encap-
sidate the viral genome to form a virion (Krupovic and Koonin 2.2 Total RNA extraction and RNA sequencing
2017). However, many positive-sense single-stranded RNA vi- Total RNA of each isolate was extracted from mycelia using an
ruses are capsidless, including Narnaviridae, Mitoviridae, RNAiso Plus kit (TaKaRa, China) according to the manufac-
Hypoviridae, Endornaviridae, Umbravirus, among several others
turer’s instructions and then treated with DNase I. The concen-

Downloaded from https://academic.oup.com/ve/article/7/1/veab032/6206434 by Universidade Federal de Viçosa user on 25 March 2022


(Dolja et al. 2012).
tration of each RNA extraction was measured using a Nanodrop
Mycoviruses replicate in all lineages within kingdom Fungi
2000 spectrophotometer (Thermo Fisher Scientific, USA). Total
and have been described in phytopathogenic, entomopatho-
RNA was stored at 80  C until being used for RNA-Seq analysis.
genic, medical, and edible fungi (Pearson et al. 2009; Xie and
Equal amounts of total RNA from 100 isolates collected annually
Jiang 2014; Ghabrial et al. 2015; Kotta-Loizou and Coutts 2017).
were pooled in a single library according to the collection year.
Most mycovirus infections cause few or negligible symptoms
Three RNA libraries were generated and used for sequence
detectable in the host (Ghabrial and Suzuki 2009). However, a
portion of the mycoviruses can reduce fungal growth, sporula- analysis. All library preparations and sequencing steps were
tion, virulence, and mycotoxin production and cause fruiting performed by GENEWIZ Inc. (China). rRNA was depleted from
bodies to deform (Xie and Jiang 2014). Mycoviruses have also total RNA using the Illumina Ribo Zero rRNA removal Kit (Plant
been reported to enhance abiotic stress tolerance, pathogenic- Leaf). Three libraries were constructed according to the manu-
ity, or the fitness of their fungal host (Ahn and Lee 2001; facturer’s protocol (NEBNext Ultra Directional RNA Library Prep
Marquez et al. 2007; Ozkan and Coutts 2015). In practical use, Kit for Illumina). Paired-end (150 bp) sequencing of the three
some of the mycoviruses known to induce hypovirulence have cDNA libraries was performed on the HiSeq 2500 platform
been developed as virocontrol agents against plant fungal dis- (Illumina, USA). To obtain the 5’ and 3’ terminal sequences of
eases (Nuss 2005; Ghabrial et al. 2015). the viral genome, the previously described method was used
Sclerotinia sclerotiorum is a devastating phytopathogenic fun- (Potgieter et al. 2009). Primers used in this study are listed in
gus with a worldwide distribution and can infect more than 700 Supplementary Table S1.
species of plants, including important crops and numerous
weeds (Farr and Rossman 2020). Sclerotinia stem rot caused by
S. sclerotiorum is one of the most serious diseases of annual 2.3 Virus-related sequence assembly and annotation
rapeseed, potentially resulting in yield losses as great as 80% Adapter sequences and low-quality reads were removed from
and significantly reducing oil content and quality (Sharma et al. the raw reads of each library using the Trimmomatic program
2015). Sclerotinia sclerotiorum harbors a great diversity with (version 0.36) (Bolger et al. 2014) with default parameter set-
eighty-nine mycoviruses documented in the GenBank database
tings. The remaining reads were mapped to the S. sclerotiorum
(update date: 26 September 2020), some of which have great po-
genome (http://fungi.ensembl.org/Sclerotinia_sclerotiorum_
tential to be developed as virocontrol agents. For instance, a cir-
1980_uf_70_gca_001857865/Info/Index) using HISAT2 (version
cular single-stranded DNA virus that confers hypovirulence has
2.1.0) (Kim et al. 2015). Unmapped reads were extracted from
been shown to manage sclerotinia disease effectively under
the BAM file using SAMtools (version: 1.9) (Li et al. 2009) and
field conditions (Yu et al. 2010; Yu et al. 2013; Zhang et al. 2020).
were then assembled de novo using Trinity (version 2.5.0) (Haas
In this study, to investigate the spread and prevalence of
et al. 2013) and SPAdes (version 3.11.1) (Bankevich et al. 2012).
mycoviruses in S. sclerotiorum, we focused on mycovirus biodi-
versity and the annual variation within a single field for three After the removal of all contigs shorter than 1,000 bp, the
years at the end of growing seasons. The results suggest that remaining contigs were filtered for redundancy at 90% nucleo-
even within a small geographical area, the S. sclerotiorum virome tide identity using CD-HIT (Fu et al. 2012). To identify virus-re-
consists of a highly variable mycovirus community that not lated contigs, the assembled contigs were compared against the
only includes a stably maintained core population under field NCBI nonredundant protein database using DIAMOND BLASTX
conditions but also harbors a dynamic portion of the mycovirus (version 0.9.10) (Buchfink et al. 2015). We set the e-value below
compositions. We aimed to define novel mycoviruses in explor- 1  105 to maintain high sensitivity and a low false-positive
ing the diversity and evolution of viruses detected from a field rate. Contigs matched to the same reference sequence were
population of S. sclerotiorum, as well as initiating a platform for merged into longer contigs using DNAMAN software (version
continuously monitoring fungus–mycovirus dynamics in natu- 8.0; Lynnon Biosoft, Quebec, Canada). Nucleotide sequences, po-
ral fungal communities. tential ORFs, and deduced amino acid sequences were analyzed
using DNAMAN.

2. Materials and methods


2.1 Fungus isolation and purification 2.4 Quantification of relative transcript abundances

From the pith of rapeseed stems in the late stage of the disease, We used the assembled contigs and S. sclerotiorum transcripts as
sclerotia were collected annually for three years from 2016 to templates for mapping with the program Bowtie2 (Langmead
2018. More than 100 sclerotia were collected each year from dis- and Salzberg 2012). Next, we measured the abundance of these
eased stems (one sclerotium from each diseased stem) that sequences as the number of transcripts per million (TPM) within
were randomly sampled from a single field (approximately each sequencing library. The relative abundance of each viral
667 m2) located in Wuxue City (N29 590 3.3600 , E115 270 58.8400 ), transcript was calculated as the TPM using RSEM (Li et al. 2011)
Hubei Province, China. All sclerotia were isolated and purified and visualized in a heat map by TBtools (Chen et al. 2020).
J. Jia et al. | 3

2.5 Phylogenetic analysis of mycoviruses 3.2 Novel positive-sense single-stranded RNA viruses
To determine the evolutionary relationships among the newly In this study, the forty-three putative viruses encoded by fifty-
identified viruses, the RdRp encoded by the putative mycovi- two contigs as diverse as previously characterized þssRNA vi-
ruses was subjected to BLASTp analysis with the NCBI-nr data- ruses (Supplementary Table S2), signifying that S. sclerotiorum
base. The core conserved domain of RdRp was aligned with does harbor abundant þssRNA mycoviruses with great diversity
MAFFT (version 7.427) using the E-INS-i settings (Nakamura regardless of the geographical origins.
et al. 2018). All alignments were trimmed with trimAl (v1.4) Four novel multi-segmented narnaviruses were identified,
(Capella-Gutierrez et al. 2009) to remove low-quality regions which were temporarily named as Sclerotinia sclerotiorum nar-

Downloaded from https://academic.oup.com/ve/article/7/1/veab032/6206434 by Universidade Federal de Viçosa user on 25 March 2022


through heuristic selection under the automatic method (-auto- navirus 1 (SsNV1), SsNV2, SsNV3, and SsNV4, representing the
mated1) based on similarity statistics. The maximum likelihood first discovery of narnaviruses in S. sclerotiorum. RT-PCR analysis
(ML) phylogenetic trees were constructed using the IQ-TREE of the cDNA of each isolate included in the NGS sample 2017SS
(version 1.6.11) (Nguyen et al. 2015) with 1,000 bootstrap repli- showed that all segments of the four segmented narna-like vi-
cates and the best-fit amino acid substitution models were ruses coexisted in the corresponding fungal isolate
identified using ModelFinder (Kalyaanamoorthy et al. 2017). (Supplementary Fig. S2). The 5’ and 3’ terminal sequences of the
Nodes supported by a bootstrap value below 70% were collapsed four multi-segmented narnaviruses were obtained. The 5’ and
in TreeGraph 2 (Stöver and Müller 2010). Phylogenetic trees 3’-terminal 40 nt were highly conserved in the multi-segments
were visualized in FigTree v1.4.3 (http://tree.bio.ed.ac.uk/soft of each narnavirus (Supplementary Fig. S3A). SsNV1 and SsNV2,
ware/figtree/). The percent identity matrix among protein-cod- respectively, have quad- and tri-segmented genomes (Fig. 1A).
ing sequences was generated using Clustal Omega for multiple Interestingly, complete RdRps of SsNV1 and SsNV2 were
sequence alignment (Li et al. 2015). encoded by two different RNA segments. RNA1 of SsNV1 and
SsNV2 harbored the G, F, A, and B motifs of the RdRp at the C-
terminus, whereas RNA2 of these two viruses contained the C,
D, and E motifs at its N-terminus (Fig. 1A and Supplementary
2.6 Availability of data and materials
Fig. S3B). RNA1 of SsNV1 and SsNV2 encoded proteins share less
The raw sequence reads from the metagenomic libraries are than 62% identity to other narnaviruses, while proteins encoded
available at the NCBI Sequence Read Archive (SRA) database un- by RNA2, RNA3, and RNA4 of these two viruses share no signifi-
der BioProject accession number PRJNA598316. The sequences cant similarity with known sequences in either the NCBI nr
reported in this research have been deposited in GenBank data- database or in the Pfam domain database. In addition, the other
bases, and the assembled contig sequences are available from seventeen narnaviruses with multi-segmented genomes were
GenBank databases under the accession numbers indicated in identified from public data (NCBI SRA and TSA database) (Fig. 1B
Supplementary Table S2. All virus nucleotide sequences (Fasta and Supplementary Table S3). The phylogenetic tree suggested
format), the unaligned and aligned datasets used in the phylo- that these divided RdRp narnaviruses formed an independent
genetic analyses (Fasta format), and the phylogenetic trees phylogenetic cluster, which was significantly distant from other
(Newick format) that were generated in this work are available non-segmented narnaviruses (Fig. 1B). Therefore, we propose a
at the Figshare website (DOI: 10.6084/m9.figshare.11474709). new family, Polynarnaviridae, to accommodate this virus clade.
SsNV3 and SsNV4 have bi-segmented genomes (Fig. 1A). The
RNA1 segment contains an ORF encoding an essential RdRp,
and the RNA2 fragment contains an ORF encoding no similarity
3. Results with the known proteins in the NCBI nr database. BLASTp
3.1 Metatranscriptomic identification of mycoviruses analysis showed that the protein with the top hit for ORF1 of
from S. sclerotiorum in a single rapeseed field SsNV3 was RdRp from Plasmopara viticola lesion-associated
orfanplasmovirus 4 (PvLaOrfPlV4) (33.75% identity), whereas
We initially analyzed the growth rate, colony morphology, and
that for ORF1 of SsNV4 was RdRp from PvLaOrfPlV5 (72.16%
pathogenicity for the 100 isolates of S. sclerotiorum collected in identity) (Supplementary Table S2). However, different from the
May 2017, and the results showed that these isolates were sig- genomes of PvLaOrfPlV (1–5) that were reported to contain a sin-
nificantly different in these three biological characteristics gle RNA segment, SsNV3 and SsNV4 have two RNA segments.
(Supplementary Fig. S1). Nearly 50% of the isolates grew fast Interestingly, the proteins encoded by RNA2 of SsNV3 are most
and had strong pathogenicity, while some isolates grew slowly similar to coat proteins of the known viruses, based on the pre-
with reduced pathogenicity (Supplementary Fig. S1C and D). diction by I-TASSER and Phyre2 (Supplementary Fig. S4).
These phenotypic differences indicate that there may be abun- Therefore, we speculate that the genomes of SsNV3 and SsNV4
dant mycoviruses in the selected field site. To understand the may not be naked, but encapsulated in virions. Phylogenetic
interannual dynamics in diversity and abundance of mycovi- analysis demonstrated that SsNV3 and SsNV4 grouped as a
ruses in this field, a total of 300 isolates were collected and used cluster with those unsegmented narnaviruses, but clustered
to construct three metatranscriptomic sequencing libraries into a separate branch with PvLaOrfPlV (1–5) (Fig. 1B).
according to the collection year. Total coverage of more than Twelve novel mycoviruses share RdRp identity with mem-
100 million reads passing the filtering criteria was obtained for bers of the Botourmiaviridae family, ranging from 23% to 82%
each library. For simplicity, we named the three libraries (Supplementary Fig. S5A). The obtained genomes ranged from
2016SS, 2017SS, and 2018SS based on the collection year. Based 1.99 to 3.85 kb in length, and each genome contained a single
on the analysis of the assembled contigs, a total of sixty-eight complete ORF (Supplementary Fig. S5B). Phylogenetic analysis
putative mycoviruses with nearly complete genomes (contain- suggested that twelve botourmiaviruses clustered into three
ing complete ORFs) or partial genomes were identified well-supported clades, among which five botourmiaviruses be-
(Supplementary Table S2). long to the genus Penoulivirus, four belong to the genus
4 | Virus Evolution, 2021, Vol. 7, No. 1

2568 1 3453
A RNA1 5' G F A B 3'
RNA1 5' RdRp 3'
42 2513

SsNV3
2285 8 3271
RNA2 5' C D E 3' 1 3100
SsNV1
50 2188
173 523 664 1020 RNA2 5' Putative coat protein 3'
8 2874
1168
RNA3 5' 3' 1 3105
42 1088
257 505 RNA1 5' RdRp 3'

SsNV4
7 2970
899
RNA4 5' 3' 1 2477

Downloaded from https://academic.oup.com/ve/article/7/1/veab032/6206434 by Universidade Federal de Viçosa user on 25 March 2022


43 768
RNA2 5' Putative coat protein 3'
55 318
2384 7 2358
RNA1 5' G F A B 3'
38 2302
SsNV2

2259
RNA2 5' C D E 3'
50 2173
730 1026
Nar
nav

PvNlV1 10
PVaNarna14
PVaNarna13
PVaNarna 30

PVaNarna9
PVaNa a11
3'

PVaN
RNA3 5' irid

PVa eyNLV1
PVaN

PVaNarna

PVa Narna 25
PVa arna38
1099

PvL aOrfP 4
ae
PVa arna3 7
56 805

BNlV22

Pv aOrfP lV2
PVa arna 4

BNlV23

PvL Narna2
WiNV2
NpNV1

Leps V1

Pv LaOrf lV1
PVa Narna 35

PiRV-4
PV

Pv LaOr PlV3
Narn

arn
N

PserN
PV Narn na36

arna3 2

Ss NV4 rfPlV 4
rna
N

Ss LaO fPlV
PV aNar NV1 0

5
a
N
PV Gn a2 1

aN

26
a3
a
arn 12
PV Hp SP a7

S N 20 a1
3

B
1
aN afN V1

a
3

RmcNV arn
S aN 3
a

P NlV V- S
a

FO NV

Sc VaN 4 20S
arn V
PV
Om ar V

S 21
aN PvN V2 1

2 3 rna
NV a
S
100
n 1
Af faN

PV 23 40
M
uN V

H
aN laN V- na
N ar 1
100
100
Af arn V3 m N V
e

R Va nN 1
100
ida

uN a6
100

P we NV
10

F
10

100 1 0 0 0
PV N pNV V1

10
0

B lun V2
0
vir

100

aN pN 2
100

100 B NL 1
O vNV 1

0
ar V 2
95
rna

10
PV
10
100

n
aN RsN a5 C xNV 1

0
1
0
10

C NLV rna4

10
arn V1
0
yna

100
95

PV MfaN a19 O aNa V1


10
0

100

P V uN L n a 4 2
10

a 10 1 0
PV Narn V3
0

10 0
Pol

0 0
Lh a N a r a 1 5
100

aN
a rn a 3
0
10
PV aNarn a43
10
97

0 10
AtN a 4 10 0
74

0
PV aNarn 23
10

0 10

10
97

Hla V 0
0

NV21
80
PV Narna 2
98

1 00

H
P V a N a rn a 2
3
PVa laNV1
10

0 99 9

0
0

99 0 10
Narn PVa Narn a 18
96

95 10
0
0
10
100

10

PVaN a2 10 10
1 100 PVa arna17
0 10

arn a 10000
0
10
0
SsNV 1 PVaN 1
10

1 00
99

10 0
Mm a N 1 100 99
10
0
10 0 FpNV rna16
V
MmaNV 3
100
1 74
PVaNa
10 0
SsOV5
10 0
99

MmaNV2
100 77
SsNV2 70 10 0 99
100
100 SsOV4
100 PVaOurmia91

Penoulivirus
CtNV1 100 99 100
76
HpafNV2 100
PmOLV1
98 80 84 CuOulV2
MfaNV2 100
100 84 SsOV7
100
100 SsOV6
100 100 AnOlV1
10 0

0
92 PlRV1
10 0 10 10
94 0 PoOLV
idae 98 7 79 100 100 SsOV9
Levivir
100 79 6
96 100 99 93
10
0 10
0 SsOV
98 10 10 10 74 SsOV 8

Boto
1 00 99 0 10 0
0
B631 SsO 13
85

10
V1-N
0 0
73
10

SsO V12
0

1
CpM DapiMV 1
10

99
0

SsO V3

Boto
rMV
10

80

u
100

Ama uMV1
0

EnO V2
0

r
10
10

99
92

mia
85

Bev aMV1
10 0

BO ulV1
10
10

100
0

uliv
Ss LV
0

Cas qMV17
96
100

10
100

Ss OV11
vir
0
0

C V2
10
99
10

100

M 9 Ss OV1
92

iru
100

Ss MV2 8
100
0

99

ida
0

10

99
10

Ss OV 0
0

Ss MV2 1
10

100

100
100 0

100

Sla OV1 14
s
100
10

100

Ss aMV 3
99
0

99
100

C O
100
10

A MV 3
99

S RM urV
S
100
100
100

Fp MV 5
0

cle

Ss sOV V 1-1
100
10

Bc V3 1 P O 1
ro

M V
Ss cM V4 3 C c O V1 6
M cO LV 5
u

B pM V
l

O ul 1
i

F sM
Ss Ss V3- V2

vir

LV V1
O C
M MV WX

S
M nM

1
uM V
Ep sVC 7
Ss V7-W 12

us
C sV V
Ss S

S V X

V
W aN
Ss sMV 20

M
Tu N l V 3 1
Ss V3 7

ag
H NlV 36
0
M

W MV
Ss V37

ou
Ss MV1
MV 6

Sn MV4
M

SsM 11
Ss MV

liv
Bc V4
AbMV14
M

SsM V39
SN V1

Ou
SsM V2
CrM lV7

iru
FpM V1
HNlV V1

CfM V38
SsM 25

rm
CcMV 32

SsM
SsMV a

SsM 5

s
15

SsMV 8
7

SsMV
1

SsMV2

BcMV2
SsMV9
SsMV19

CfMV1
V

SsMV33

ia
AbMV1
AaMV1
SsMV1

SsMV8 FpMV1

in

vi
V1

Mi
ru
ve
Ou bra

tov
1

s
rte

0.5 irid
r m te

ae
ia- vir
lik us
e es

Fungi Invertebrates Plants Bacteria Oomycetes Marine microorganisms

Figure 1. Genome organizations of narna-like viruses and phylogenetic relationships of the members in the ‘Narna-Mito-Botourmia’ clade. (A) Genome structure mod-
els for the narna-like viruses identified in this study. The colored boxes indicate hypothetical ORFs. (B) The ML tree was constructed based on the multiple amino acid
sequence alignment of the core conserved domain of RdRps using IQ-TREE with the best-fit model ‘Blosum62þFþR7’. The first identified viruses are marked by red col-
ors, and the identified polynarnaviruses from NCBI SRA or TSA database are marked by blue colors. Sequence information of all selected viruses were supplied in
Supplementary Table S4. The numbers next to each branch represent the bootstrap support based on 1,000 replicates. All branch lengths are drawn to a scale of amino
acid substitutions per site. Host taxa are shown by circles delineated in different colors. The abbreviation ‘WX’ represents Wuxue (WX) city, which was used to distin-
guish the identified viruses in this study from the previously reported mycoviruses with more than 90% amino acid identity.
J. Jia et al. | 5

Botoulivirus, and another three belong to the genus Scleroulivirus Solemoviridae, Ambiguiviridae, Endornaviridae, Botybirnavirus, and
(Fig. 1B). one unassigned virus species. Due to space limitations, these
We identified a novel virus belonging to the family mycoviruses are described in Supplementary Text S1.
Fusariviridae, tentatively named Sclerotinia sclerotiorum fusar-
ivirus 2 (SsFV2). The assembled genome of SsFV2 is 6,281 nt in 3.3 Novel negative-sense single-stranded RNA viruses
length and encodes two nonoverlapping ORFs (Supplementary
Fig. S6A). ORF1 encodes a polyprotein with conserved RdRp and We identified five -ssRNA mycoviruses belonging to the family
helicase (Hel) domains showing 51.29% identity with previously Mymonaviridae. The sequences of these five mycoviruses ranged
described Rutstroemia firma fusarivirus 1. ORF2 encodes a hy- from 9,623 to 9,982 nt, suggesting that the complete or nearly

Downloaded from https://academic.oup.com/ve/article/7/1/veab032/6206434 by Universidade Federal de Viçosa user on 25 March 2022


pothetical protein of unknown function. Phylogenetic analysis complete genomes of all these viruses were obtained based on
based on the conserved RdRps showed that SsFV2 and other the deep-sequencing data. The L proteins of Sclerotinia sclero-
members of the Fusariviridae family are divided into three tiorum negative-stranded RNA virus 1 (SsNSRV1-WX) and
branches with good support (Supplementary Fig. S6B). For this SsNSRV2-WX shared 100% and 99% identity with SsNSRV1
reason, we propose the division of this family into three genera: (KJ186782.1) and SsNSRV2 (KP900931.1), respectively
Alpha-, Beta-, and Gammafusarivirus, among which SsFV2 (Supplementary Fig. S10A). Similar to that of other members of
belongs to the Alphafusarivirus genus. Mymonaviridae, the genome of SsNSRV9 contained five nonover-
One mycovirus, Sclerotinia sclerotiorum virga-like virus 1 lapping ORFs (ORF I–V) arranged linearly in the viral genome
(SsVV1), was identified. The SsVV1 genome consists of two RNA (Supplementary Fig. S10B). The protein encoded by ORF IV
segments, designated as RNA1 (1,800 nt) and RNA2 (1,878 nt), in SsNSRV9 shared 56% identity with the L protein in SsNSRV1
which encode RdRp and methyltransferase (Met), respectively and SsNSRV3 (Supplementary Fig. S10A). In addition, a con-
(Supplementary Fig. S7A). The predicted amino acid sequence served gene-junction sequence (30 -UAUU(U/
of RNA1 showed 44% identity with the RdRp of Agaricus bispo- A)AAUAAAACUUAGGAA-50 ) was found downstream of each
rus virus 16 (AbV16; KY357502). The Met encoded by RNA2 of ORF (Supplementary Fig. S10D), which is a common feature in
SsVV1 had an identity of 29% with AbV16. AbV16 is a proposed mononegaviruses (Whelan et al. 2004). Notably, SsNSRV10
member of the Mycovirgaviridae family (Nerva et al. 2019), and SsNSRV11 contained four ORFs (ORF I–IV) (Supplementary
whose members exhibit four RNA segments, containing a single Fig. S10B), but there was only a transcription start signal with
ORF: segment 1 encoding RdRp, segment 2 encoding Met, and no stop signal upstream of ORF I and only a transcription
segments 3 and 4 encoding proteins of unknown function stop signal with no start signal downstream of ORF IV
(Deakin et al. 2017). Although both RNA1 and RNA2 of SsVV1 (Supplementary Fig. S10D); therefore, we believe that these two
were similar to those of AbV16, we failed to detect the other two mycoviruses may have only four ORFs. However, SsNSRV9 is
segments corresponding to those of AbV16 from our data. similar to the previously reported SsNSRV1 in that there is no
Phylogenetic analysis revealed that SsVV1 clusters in a clade transcription initiation signal in their 3’-UTR (Liu et al. 2014).
with AbV16 along with several other viruses recently discovered Phylogenetic analysis of the L protein confirmed that SsNSRV10
in soil samples and fungal transcriptome data (Supplementary and SsNSRV11 formed an independent evolutionary branch,
Fig. S7B). while SsNSRV9 was clustered with SsNSRV1 and related viruses
We also identified three mycoviruses belonging to the (Fig. 2).
Deltaflexiviridae family, tentatively named Sclerotinia sclerotio- Sclerotinia sclerotiorum ophiovirus-like virus 1 (SsOpLV1)
rum deltaflexivirus 1-WX (SsDFV1-WX), SsDFV2-WX, and with 7,817 nt in length shows the highest similarity to Fusarium
SsDFV3. The replicases of SsDFV1-WX and SsDFV2-WX shared poae negative-stranded virus 1 within the recently proposed
99% and 98% identity with SsDFV1 (NC_038977) and SsDFV2 Mycoaspiviridae family (Chiapello et al. 2020). SsOpLV1
(NC_040649), respectively (Supplementary Fig. S8). The assem- contained two linearly arranged nonoverlapping ORFs
bled genome of SsDFV3 consisted of 7,375 nt with two predicted (Supplementary Fig. S10C). The ORF1 encoded a protein (143 aa)
ORFs. ORF1 encodes a putative replication-associated polypro- of unknown function, and ORF2 encoded a polyprotein with the
tein and contains three conserved domains: Met, Hel, and RdRp RdRp domain. Phylogenetic analysis based on RdRp indicated
(Supplementary Fig. S7A). A BLASTp search using the deduced that mycoophioviruses are closely related to the viruses of the
amino acid sequence showed that ORF1 has a low identity Aspiviridae family (Fig. 2).
(<40%) with the replication-associated polyprotein of members Seven novel mycoviruses related to the members of
in the Deltaflexiviridae family (Supplementary Fig. S8). Bunyavirales were identified. The genomes for the seven bunya-
Phylogenetic analysis revealed that SsDFV3 was in a well- viruses (Sclerotinia sclerotiorum bunyavirus 1-7 (SsBYV1-7))
supported clade with other deltaflexiviruses (Supplementary ranged from 6.4 to 8.0 kb, and contain a single ORF
Fig. S7B). (Supplementary Fig. S11A). The L proteins encoded by the seven
A 4470 nt contig was similar to barnavirus, tentatively bunyaviruses shared less than 26% identity with those of
named Sclerotinia sclerotiorum barnavirus 1 (SsBNV1). SsBNV1 known bunyaviruses (Supplementary Table S2). We failed to
was a monosegmented þssRNA virus with four ORFs find any additional segments related to bunyavirus M or S pro-
(Supplementary Fig. S9A). The four ORFs encoded a protein of teins in the assembled contig database. The alignment of the
unknown function, a putative serine protease domain, RdRp, conserved motifs of the L proteins encoded by the seven bunya-
and a capsid protein. The predicted RdRp shared 39% identity viruses and those in the other reported bunyaviruses indicates
with the Mushroom bacilliform virus belonging to the family that these mycoviruses contain eight conserved motifs
Barnaviridae. SsBNV1 within a well-supported clade of the (Supplementary Fig. S12), which represent highly conserved
Barnaviridae family is closely related to members of central regions of RdRps from members of the Bunyavirales order
Solemoviridae and Luteoviridae families (Supplementary Fig. S9B). (Amroun et al. 2017). The conserved endonuclease domain and
In addition to the mycoviruses mentioned above, other motif G (Supplementary Fig. S11B and C) were also identified in
detected þssRNA viruses and dsRNA viruses included members the N-terminal of the L protein encoded by three bunyaviruses
of Mitoviridae, Gammaflexiviridae, Tymoviridae, Hypoviridae, (SsBYV5-7). However, the endonuclease domain and motif G
6 | Virus Evolution, 2021, Vol. 7, No. 1

100 Sclerotinia sclerotiorum negative-stranded RNA virus 1-WX


100 Sclerotinia sclerotiorum negative-stranded RNA virus 1 | NC_025383
99 Sclerotinia sclerotiorum negative-stranded RNA virus 3 | NC_026732
95 Sclerotinia sclerotiorum negative-stranded RNA virus 9
100 Soybean leaf-associated negative-stranded RNA virus 2 | KT598227
Fusarium graminearum negative-stranded RNA virus 1 | MF276904
100 Soybean leaf-associated negative-stranded RNA virus 1 | KT598225
100 100 Alternaria tenuissima negative-stranded RNA virus 1 | MK584852
100 Sclerotinia sclerotiorum negative-stranded RNA virus 7 | MF444285
100 Botrytis cinerea mymonavirus 1 | MH648611
82 Penicillium cairnsense negative-stranded RNA virus 1 | MK584851
Sclerotinia sclerotiorum negative-stranded RNA virus 2-WX Mymonaviridae
100
100 Sclerotinia sclerotiorum negative-stranded RNA virus 2 | KP900931
87 100 Sclerotinia sclerotiorum negative-stranded RNA virus 4 | NC_043483

Downloaded from https://academic.oup.com/ve/article/7/1/veab032/6206434 by Universidade Federal de Viçosa user on 25 March 2022


Soybean leaf-associated negative-stranded RNA virus 3 | KT598228
100 Sclerotinia sclerotiorum negative-stranded RNA virus 11
Sclerotinia sclerotiorum negative-stranded RNA virus 10
100 Soybean leaf-associated negative-stranded RNA virus 4 | KT598229
Hubei rhabdo-like virus 4 | KX884403
Lentinula edodes negative-strand RNA virus 1 | LC466007
93
94 100 Penicillium glabrum negative-stranded RNA virus 1 | MK584853
Penicillium adametzioides negative-stranded
g RNA virus 1 | MK584858
100 Rhabdoviridae
Lishi Spider Virus 2 | NC_031259 Lispiviridae
p
100 Paramyxoviridae
y
99
98
94 100 Filoviridae
100
Pneumoviridae
100 Nyamiviridae
y
100
Bornaviridae
84 Xi h
Xincheng M
Mosquito
i Vi
Virus | NC
NC_031244
031244 Xinmoviridae
Pteromalus
Pt l puparum negative-strand
ti t d RNA virusi 1 | NC_038269
NC 038269 Artoviridae
100 99 Rhi
Rhizoctonia
t i solani
l i negative-stranded
ti t d d virus
i 2 | KP900920
100 Rhizoctonia solani negative-stranded virus 3 | KP900903
100 Rhizoctonia solani negative-stranded virus 1 | KP900919 Mycoaspiviridae
100 Cladosporium cladosporioides negative-stranded RNA virus 1 | MK584856
100 100 Fusarium poae negative-stranded virus 1 | NC_030871
100
Sclerotinia sclerotiorum ophiovirus like virus 1
Aspiviridae
100
98
100 Sclerotinia sclerotiorum negative-stranded RNA virus 5 | KF913892
Alternaria tenuissima negative-stranded RNA virus 2 | MK584855
100
100 Laurel Lake virus | KX774630 Laulavirus Phenuiviridae
100 Citrus concave gum-associated virus | NC_035759 Coguvirus
100 Lentinula edodes negative-strand RNA virus 2 | LC466008
Entoleuca phenui-like virus 1 | MF375882
100 100 Sclerotinia sclerotiorum bunyavirus 5
100 Sclerotinia sclerotiorum bunyavirus 6
99 Sclerotinia sclerotiorum bunyavirus 7 proposed
93 Wuhan Spider Virus | KM817699 Mycophenuiviridae
99 Fusarium poae negative-stranded virus 2 | NC_030872
88 99 100 Penicillium roseopurpureum negative ssRNA virus 1 | MG887749
Coniothyrium
y diplodiella
p negative-stranded
g RNA virus 1 | MK584854
Leptomonas
p moramango g leishbunyavirus
y 1 | KX280012 Leishbuviridae
100
99 100 Arenaviridae
100 Hubei myriapoda
y p virus 5 | NC_033761
_ Mypoviridae
yp
100
100 Nairoviridae
90 Wuhan Millipede
p Virus 2 | KM817696 Wupedeviridae
p
100 Rhizoctonia solani negative-stranded virus 4 | KP900923
Cladosporium cladosporioides negative-stranded
g RNA virus 2 | MK584857 New group?
100 Macrophomina phaseolina negative-stranded RNA virus 1 | KP900899
Botrytis
y cinerea negative-stranded
g RNA virus 1 | NC_028466
_ New group?
100 100
97 Peribunyaviridae
y
100 98 European mountain ash ringspot-associated emaravirus | NC_013105 Fimoviridae
W li crustacean
Wenling t virus
i 9 | NC
NC_032143
032143 Cu
Cruliviridae
dae
94 Tomato spotted
T d wilt
il tospovirus
i | NC
NC_002052
002052
100
Tospoviridae
p
100
Hantaviridae
89 Phasmaviridae
100 Sclerotinia sclerotiorum bunyavirus 1 proposed
87
100 Sclerotinia sclerotiorum bunyavirus 2
100 Sclerotinia sclerotiorum bunyavirus 3 Sclerobunyaviridae
Sclerotinia sclerotiorum bunyavirus 4
Fungi Plants Vertebrates Invertebrates
0.5
Vertebrates and Invertebrates Vertebrates, Invertebrates and plants

Figure 2. ML phylogenetic analysis depicting the evolutionary relationships of the negative-stranded RNA viruses. The best-fit model for constructing the phylogenetic
tree is ‘LGþFþR8’. The name of the virus family or genus is shown on the right. This is same as that of Fig. 1.

were not detected in the L proteins encoded by the four other We therefore propose the establishment of a new family,
bunyaviruses. A phylogenetic tree was constructed using the Mycophenuiviridae, to accommodate these similar bunyavi-
core regions of the L protein. Seven sclerotinia bunyaviruses ruses that infect fungi and invertebrates (Fig. 2).
were assigned into two clades. Four bunyaviruses (SsNYV1-4)
were well supported in one phylogenetic cluster, which was sig-
3.4 Diversity of the S. sclerotiorum virome in a single
nificantly distant from the other bunyaviruses, suggesting that
rapeseed field
these four bunyaviruses represent a new virus lineage.
Therefore, we propose the establishment of a new family, We examined the proportion of mycovirus-associated reads in
Sclerobunyaviridae, to accommodate these similar bunyavi- each library, which we found to be 4.07% for 2016SS, 9.51% for
ruses. The other three bunyaviruses (SsNYV5-7) formed well- 2017SS, and 2.89% for 2018SS (Fig. 3A). The sixty-eight mycovi-
supported clades with an invertebrate virus (Wuhan spider vi- ruses we identified from these three libraries represent four ge-
rus) and several fungal bunyaviruses but were significantly phy- nome types: positive-sense single-stranded RNA (þssRNA),
logenetically distant from members of the Phenuiviridae family. negative-sense single-stranded RNA (-ssRNA), double-stranded
J. Jia et al. | 7

A D
25 Genome type
2018
5 +ssRNA
2018 4
1 −ssRNA
2017
dsRNA
33 ssDNA
2016 10
2017 8
1
0

10

20

30

40

50

60

70

80

90

100
Proportion 36
2016 6
S. sclerotiorum Viruses Others 4

Downloaded from https://academic.oup.com/ve/article/7/1/veab032/6206434 by Universidade Federal de Viçosa user on 25 March 2022


1
B ssDNA,1,1.5% 0 5 10 15 20 25 30 35 40
Number of viruses
dsRNA,11,16.2% E
Year Virus Genus Virus Family Genome Type
Fusariviridae
Fusariv
vviridae
Penoulivirus
Alphafusariviruses
Alphafu
p usarivirusviruses
ruse
+ssRNA,43,63.2%
Botouliivirus
2016 Hbsclerovirus
Hb
Hbscle
lerovirus
i Botourm
rmiaviridae
rm
Unassigned
Unassi igned genus
gen
genu
nus
−ssRNA,13,19.1%
Sclerouulivirus Solemoviridae
Solemo
oviridae
o
Unassigned fam
Unassigned family
famii y
Betaen
ndornavirus Endorn
naviridae
n +ssRNA
Gammahypoviruses
Hypoviiiridae
Unclassified,1,2.3%
Solemoviridae,1,2.3% Mitovirrus
C Barnaviridae,1,2.3% Mitoviri
riidae
Fusariviridae,1,2.3% Betahy
Betahypoviruses
ypoviruse es
Mycovirgaviridae,1,2.3% Deltafle
exivirus Deltafle exiviridae
e aee
Botourmiaviridae,12,27.9% Tymoviridae,1,2.3% 2017 Fusagravirus
Fusagr
gravirus Ambiguiviridae
A mbigu uiviridae
u
Gammaflexiviridae,1,2.3% Mycotombusvirus
Mycoto
y ombusvirus svirus
virus Mycovirgaviridae
Mycovirgavirid
M
Mycovi
y iirgaviridae
rgaviridae
Mycovirgavirus
M
Mycov i
irgavirus
i Gammaflexiviridae
Gamm
G aflexiviri
fl i i id
daae
ae
Ambiguiviridae,2,4.7% Mycoflexivirus
Mycofle
M flexivirus
i i Fusagraviridae
Fusagr rraviridae
dae
Barnavirus
Barnav
B virus
i Barnaviridae
Barnav
B vviridae
i id e
Polynarnaviridae,2,4.7% Mycotymovirus
Mycoty
y ymovirus Tymoviridae
Tymovi
y iiridae
Narnaviridae
Narnav vviridae
dsRNA
Mycobunyavius
Mycob unyavius
Narnaviridae,2,4.7% Narnavirus
Narnav virus Botybirrrnaviridae
e
Botybirrnavirus
Mycobu
y unyaviridae
u yavirida
Deltaflexiviridae,3,7% Mycophenuivirus
Mycophenuivirus
us Polynarnaviridae
Polyna rnaviridae -ss
ss
ssRNA
Mitoviridae,7,16.3% Polyna
y arnavirus Mycophenuivirid
Mycophhenuiviridae
h ridae
dae
Endornaviridae,4,9.3% 2018
Sclerottimonavirus Mymon
naviridae
n
Hypoviridae,4,9.3% G ycircularvirus
Gemycircularvirus
Gemyc i l i Genom
Genomoviridae
moviridae
m e
Mycoaspivirus
Mycoa spivirus Mycoaspiviridae
Mycoas
sspivirida ssDNA

Figure 3. The diversity of mycoviruses detected in S. sclerotiorum. (A) Proportion of each taxonomic category in each library based on read counts. (B) The percentage
of each virus genome type. (C) The percentage of each positive, single-stranded RNA virus at the family level. (D) Mycoviruses belonging to different genome types
presented in each library. (E) Sankey diagram showing the compositions of mycovirome from different years of S. sclerotiorum field isolates.

RNA (dsRNA), and single-stranded DNA (ssDNA) (Fig. 3B). Botourmiaviridae and Mitoviridae families were most prevalent,
Among these sixty-eight mycoviruses, the amino acid sequen- accounting for up to 44.2% (Fig. 3C). The eleven identified
ces of twenty-two mycoviruses showed high identity (90%) to dsRNA mycoviruses included seven botybirnaviruses and four
previously reported viruses, twenty-eight mycoviruses repre- fusagraviruses (Supplementary Fig. S13). We also identified an
sented novel mycoviruses sharing low amino acid sequence ssDNA mycovirus that shared 100% identity to Sclerotinia scle-
identity (<70%) with previously recognized viruses, and eigh- rotiorum hypovirulence-associated DNA virus 1 (SsHADV-1) be-
teen mycoviruses had 70% and <90% amino acid identity with longing to the Genomoviridae family. Finally, the mycoviruses
previously recognized viruses (Supplementary Table S2). The with -ssRNA genomes were classified into three orders:
characteristics of all identified mycoviruses, including their Bunyavirales (seven viruses), Mononegavirales (five viruses), and
contig length and best hit in the NCBI-nr database, are summa-
Serpentovirales (one virus). These results suggest that mycovi-
rized in Supplementary Table S2. Based on the amino acid
ruses present in S. sclerotiorum are diverse, even though all iso-
sequences of the replicase encoded by the assembled contigs,
lates originated from a single rapeseed field.
all identified mycoviruses were classified. Nineteen of sixty-
eight (27.9%) mycoviruses had a lower identity (<45%) than pre-
viously characterized viruses, and eleven of these nineteen 3.5 Interannual dynamics of members in the
could not be placed into any established family. At the genus S. sclerotiorum virome
level, thirty-eight mycoviruses could be assigned to eleven
established genera, while the remaining thirty mycoviruses The composition of virome was different among the three
could be assigned to thirteen proposed genera and one unas- libraries. The list of putative mycoviruses was summarized for
signed genus. each library, and we found that 47, 52, and 35 different mycovi-
þssRNA mycoviruses were the most diverse class within the ruses were detected in the 2016SS, 2017SS, and 2018SS libraries,
three libraries, compared to other types (Fig. 3B). Forty-three respectively (Fig. 3D). In all three libraries, the number of
mycoviruses were þssRNA viruses, accounting for up to 63.2% þssRNA mycoviruses was greater than that of -ssRNA mycovi-
of the identified viruses, suggesting that S. sclerotiorum ruses. The number of dsRNA mycoviruses was less than that of
harbors abundant þssRNA mycoviruses with high diversity -ssRNA mycoviruses, and the number of ssDNA mycoviruses
(Fig. 3B). Among the þssRNA mycoviruses, members of the was the lowest, with only one of these viruses found in all
8 | Virus Evolution, 2021, Vol. 7, No. 1

libraries (Fig. 3D). The libraries and the taxonomy of mycovi- the Botybirnavirus genus, and one ssDNA mycovirus in the
ruses were summarized in a Sankey diagram (Fig. 3E). Gemycircularvirus genus (Supplementary Table S2).
Mycovirus occurrence changed within sampling times as fol- The relative abundances of the identified viral families
lows: 35.3% of mycoviruses were detected in all three years, showed differences among the years. Mycoviruses within the
25.8% were detected twice, and 39.4% were detected once. More Mitoviridae family were the most abundant, and their TPM
than 60% of the identified dsRNA mycoviruses and -ssRNA values accounted for more than 63.4% of all mycovirus TPM val-
mycoviruses were detected once, suggesting that these mycovi- ues (Fig. 4B). Botourmiaviruses were the second most common
ruses may have difficulty spreading widely in S. sclerotiorum or after mitoviruses in the 2016SS library, accounting for 23.2% of
that they may be easily eliminated. In total, twenty-four myco- the mycovirus TPM values, while the family accounted for only

Downloaded from https://academic.oup.com/ve/article/7/1/veab032/6206434 by Universidade Federal de Viçosa user on 25 March 2022


viruses were found each year, accounting for 35.3% of all identi- 2.0% in the 2018SS library. In the 2017SS library, the abundance
fied viruses (Fig. 4A), suggesting that these mycoviruses
levels of mycoviruses within the Botybirnavirus genus and
commonly exist in the population of S. sclerotiorum and are po-
Botourmiaviridae family were lower than the abundance of
tential core mycoviruses in the virome of S. sclerotiorum. Among
mycoviruses belonging to the Mitoviridae family, accounting for
these twenty-four mycoviruses, nineteen were þssRNA mycovi-
12.4% and 6.8% of the TPM values, respectively. In the 2018SS li-
ruses, including five mitoviruses, five botourmiaviruses, three
brary, the abundance of mycoviruses within the Mymonaviridae
hypoviruses, two deltaflexiviruses, a narnaviruses, a gamma-
family accounted for more than 17.3% of the total abundance,
flexivirus, a tymovirus, and a betaendornavirus. Additionally,
there are five other mycoviruses, including three -ssRNA myco- while mycoviruses in this family accounted for less than 2.0% in
viruses in the Sclerotimonavirus genus, one dsRNA mycovirus in the other two libraries. A heat map of the relative abundances
of all mycoviruses identified in the different libraries is pre-
sented in Fig. 4C. As shown in the heat map, almost all the

A C 100 SsBV1
2016 2017 Not detected 98 SsBV2 Sclerobunyaviridae
SsBV3
1.2 100 SsBV4
98 SsBV5
0.9 SsBV6 Mycophenuiviridae
100 SsBV7
9 0.6 76 SsNSRV11
9 15 95 SsNSRV10
0.3 SsNSRV2-WX Mymonaviridae
SsNSRV1-WX
0.0 95 99 SsNSRV9
24 SsOpLV1 Mycoaspiviridae
-0.3 92 SsGFV1 Gammaflexiviridae
5 4 92 SsMTV1-WX Tymoviridae
-0.6 SsULV3-WX1 Ambiguiviridae
71 98 100 SsULV3-WX2
-0.9 94 SsDFV1-WX
SsDFV3 Deltaflexiviridae
2 -1.2 100 SsDFV2-WX
99 SsEV8
86 100 Endornaviridae
SsEV1-WX
80 SsEV9
SsBNV1 Barnaviridae
2018 88 HuSRV1-WX Solemoviridae
85
SsVV1 Mycovirgaviridae
B 94 SsBRV4
100 Family/Genus 99 SsBRV1-WX
SsBRV5-WX1 Botybirnavirus
100 96 SsBRV5-WX2
Mycoaspiviridae 84 SsBRV3-WX
Mycovirgaviridae SsBRV2-WX
98 93 SsFGV1
Narnaviridae SsFGV2 Fusagraviridae
Solemoviridae 100 SsNsV-L-WX1
75 Fusariviridae SsNsV-L-WX2
97 SsHV8-WX1
Relative abundance (%)

Unclassified 99 SsHV8-WX2 Hypoviridae


Tymoviridae SsHV2-WX
99 SsHV7
Mycophenuiviridae 97 SsFV2 Fusariviridae
Endornaviridae SsOV10
100 SsOV11
Gammaflexiviridae SsOV12
50 Sclerobunyaviridae 100 SsOV13
93 SsOV5
Deltaflexiviridae SsOV6 Botourmiaviridae
97 100 SsOV7
Barnaviridae 98 SsOV15
Fusagraviridae 99 SsOV16
Hypoviridae 89 SsOV14
100 SsOV8
Genomoviridae SsOV9
25 100 SsNV1 Polynarnaviridae
Mymonaviridae
SsNV2
Botybirnavirus 99 SsNV3 Narnaviridae
Polynarnaviridae 70 100 SsNV4
100 SsMV37
Ambiguiviridae 91 SsMV3-WX
94
Botourmiaviridae SsMV35 Mitoviridae
SsMV7-WX
0 Mitoviridae 99 100 SsMV36
2016 2017 2018 SsMV39
100 SsMV38
SsHADV1-WX Genomoviridae
2016 2017 2018

Figure 4. The abundances of mycoviruses in S. sclerotiorum from different years. (A) The number of shared mycoviruses in the virome of S. sclerotiorum in three years. (B)
The relative abundance of mycoviruses at the family level in the same year, normalized by column. (C) Heat map displaying the relative abundance of the mycoviruses
in different years according to the TPM values, normalized by row. The red columns show high abundance, and grey columns indicate not detected. Mycoviruses
detected in all three libraries are marked with green dots.
J. Jia et al. | 9

mycoviruses had changing abundances between sampling the mycovirus composition in a single field was found similar
years. to the previously reported virome, although the numbers of spe-
cies were more diverse. Moreover, most of these mycoviruses
did not exhibit any specific association to geography or plant
4. Discussion host. This result also revealed that the virome of S. sclerotiorum
Metatranscriptomics sequencing has dramatically expanded from a single crop field in the same region might not differ sig-
our understanding of virus diversity and allowed novel mycovi- nificantly from the overall virome of S. sclerotiorum sampled
ruses to be increasingly discovered in phytopathogenic fungi from other plant hosts and geographical origins. Whether
(Marzano et al. 2016; Kotta-Loizou and Coutts 2017; Mu et al. viromes in other species of fungal populations have similar

Downloaded from https://academic.oup.com/ve/article/7/1/veab032/6206434 by Universidade Federal de Viçosa user on 25 March 2022


2017; Arjona-Lopez et al. 2018; Nerva et al. 2019; Chiapello et al. results is still unclear and yet to be explored for the dynamics
2020). However, the understanding on the interannual dynam- among the plant disease severity, fungal hosts, and mycovi-
ics of mycovirus under natural conditions is limited at a popula- ruses at population levels.
tion level. In the present research, we performed viral Previous reports brought insights into the important roles
metatranscriptomics sequencing to investigate the interannual viruses play in natural ecosystems (Suttle 2007; Dell’Anno et al.
dynamics and characterize unbiased viral communities in S. scle- 2015). Understanding the temporal dynamics of viromes could
rotiorum collected from a single rapeseed-growing field for three help us determine the drivers of community stability and eco-
consecutive years. As expected, the species and number of the system function generally in nature (Rodriguez-Brito et al. 2010;
identified mycoviruses showed variation in an individual year, Arkhipova et al. 2018; Ignacio-Espinoza et al. 2019) and specifi-
but it is noteworthy that twenty-four mycoviruses detected in all cally in agricultural fields. For instance, the virus population
three years are considered to be core mycoviruses in S. sclerotio- can co-vary with prokaryotic abundance by preying on the
rum virome. Additionally, some of the mycoviruses discovered in microbes and change the microbial community composition,
this study were classified into families of Narnaviridae, and thereby modulating ecosystem functions such as carbon
Polynarnaviridae, Gammaflexiviridae, Mycovirgaviridae, Barnaviridae, and nitrogen cycling in the natural environment (Suttle 2007).
Mycoaspiviridae, Mycophenuiviridae, Sclerobunyaviridae with The temporal dynamics of several environmental viromes have
S. sclerotiorum as the host for the first time. The findings enrich been characterized in recent decades, including bioaerosol
our understanding on the diversity of mycoviruses and provided (Prussin et al. 2019), ocean (Chow and Fuhrman 2012), sphag-
important evolutionary clues. num peatlands (Ballaud et al. 2016), and sediment ecosystems
In this study, sixty-eight mycoviruses were detected in (Helton et al. 2012). In this study, we found that the mycovirus
S. sclerotiorum, and besides one unclassified mycovirus, the communities exhibited fluctuating temporal abundance, with
other sixty-seven mycoviruses were assigned into twenty fami- the viral populations in 2017 being larger than those in the other
lies, suggesting that S. sclerotiorum harbors a highly complex two years. In contrast to mycoviruses, another study analyzed
mycovirus composition, even in a single crop field. Moreover, plant viruses from six native plant species over four years in the
twenty-eight mycoviruses were previously uncharacterized, in- Nature Conservancy’s Tallgrass Prairie Preserve, Osage County,
dicating mycoviral communities in S. sclerotiorum are still poorly Oklahoma, USA. The taxonomic composition of plant viruses
covered by current databases. Two virome analyses were previ- was found to have a close relationship with the plant host spe-
ously conducted in S. sclerotinia. Twenty-eight mycoviruses cies but no clear relationship with the sampling site or year
were detected from 138 isolates of S. sclerotiorum in the USA, be- (Thapa et al. 2015). Although most mycoviruses cause latent
longing to eight families (nine genera) (Marzano et al. 2016), of infections, the mycoviruses associated with hypovirulence/
which members from seven families (eight genera) were found hypervirulence or other biological functions have been widely
in the present research. Fifty-seven mycoviruses were detected reported in various phytopathogenic or nonpathogenic fungi
from eighty-four isolates in Australia, belonging to thirteen (Marquez et al. 2007; Ghabrial et al. 2015; Kotta-Loizou and
families (fourteen genera) (Mu et al. 2017), of which members Coutts 2017; Nerva et al. 2019). Mycoviruses also can influence
within ten families (eleven genera) were detected in our sam- the plant–endophyte relationship by changing the virulence or
ples. Mitoviruses are the most prevalent in S. sclerotiorum sam- fitness of the fungal host under altered environmental condi-
ples from Australia and the USA, and the proportion of them in tions (Bao and Roossinck 2013). Bio-priming treatment of rape-
those two viromes of S. sclerotiorum are 43.9% (25/57) and 60.7% seeds with the hypovirulent strain DT-8 infected with a
(17/28), respectively. Unlike samples from the United States and mycovirus, SsHADV-1, could influence the structure and com-
Australia, botourmiaviruses were most prevalent in this virome position of overall fungal communities in Sclerotinia stem rot
survey of S. sclerotiorum from China, accounting for 17.6% (12/ (Qu et al. 2020). Therefore, we believe that mycoviruses may
68), followed by mitoviruses (7/68) and botybirnaviruses (7/68). play an essential role in fungal communities in the agricultural
Based on the percentage of genome types, our findings were ecosystem via complex fungi–mycovirus interactions. However,
similar to those of previously reported results (Marzano et al. the temporal dynamics of virome have not been explored at a
2016; Mu et al. 2017), with þssRNA viruses as the dominant pop- fungal community level. Therefore, our study represents the
ulation accounting for more than 60% of the identified mycovi- first effort in quantitatively and qualitatively describing the
ruses. S. sclerotiorum virome showed a similar mycovirus temporal dynamics of the virome in fungi. Previous research
composition despite location as S. sclerotiorum isolates from confirmed that the occurrence of sclerotinia disease was
Australia, USA, and China are from different continents sepa- affected by many factors including temperature, humidity,
rated by oceans. Therefore, we boldly speculated that the inter- and host resistance (Sharma et al. 2015). However, beyond
action system between S. sclerotiorum and its core virome had abiotic and host factors, we speculated that the virulence of the
been initiated before continental plate separation. S. sclerotiorum population taken as a whole was also greatly
The S. sclerotiorum isolates of the USA and Australia were col- affected by mycoviruses for the following reasons. First, the
lected from different geographical regions and plant hosts, isolates of S. sclerotiorum collected from a single crop field
while strains of the Chinese group were isolated from a smaller showed significant virulence variation (Attanayake et al. 2013;
annual rapeseed-growing field. However, as mentioned above, Abán et al. 2018), and a wide variety of mycoviruses have been
10 | Virus Evolution, 2021, Vol. 7, No. 1

discovered in S. sclerotiorum (Jiang et al. 2013; Marzano et al. the earmarked fund for China Agriculture Research System
2016; Mu et al. 2017). Secondly, some mycoviruses could not (CARS-12).
only confer hypovirulence on S. sclerotiorum but also have the
potential to convert S. sclerotiorum into an endophyte and en-
hance the resistance of rapeseed against the phytopathogens,
Supplementary data
for example Botrytis cinerea and S. sclerotiorum (Zhang et al. 2020). Supplementary data are available at Virus Evolution online.
Third, biopriming with hypovirulent strain DT-8 could reduce
the total abundance of the potential phytopathogenic fungi (Qu Conflict of interest: None declared.
et al. 2020). Therefore, we speculate that the interannual dy-

Downloaded from https://academic.oup.com/ve/article/7/1/veab032/6206434 by Universidade Federal de Viçosa user on 25 March 2022


namics of virome in S. sclerotiorum may be the hidden driver in References
shaping the fungal community structure that then may reduce
Abán, C. L. et al. (2018) ‘Molecular, Morphological and Pathogenic
the occurrence of diseases caused by other fungal pathogens.
Diversity of Sclerotinia sclerotiorum Isolates from Common Bean
It is generally thought that the RdRp of all RNA viruses is
(Phaseolus vulgaris) Fields in Argentina’, Plant Pathology, 67:
encoded by a single segment of gene and the phylogeny of RNA
1740–8.
viruses is fully characterized based on the viral RdRp
Ahn, I.-P., and Lee, Y.-H. (2001) ‘A Viral Double-Stranded RNA up
(te Velthuis 2014; Wolf et al. 2018). We found the RdRps of two
Regulates the Fungal Virulence of Nectria Radicicola’, Molecular
novel multi-segmented polynarnaviruses encoded by two dif-
Plant-Microbe Interactions: Mpmi, 14: 496–507.
ferent segments and this similar result was recently reported in
Amroun, A. et al. (2017) ‘Bunyaviridae RdRps: Structure, Motifs,
Aspergillus fumigatus, Magnaporthe oryzae, and Oidiodendron maius
and RNA Synthesis Machinery’, Critical Reviews in Microbiology,
(Chiba et al. 2020; Sutela et al. 2020). By analyzing NCBI SRA and
TSA data, we found that seventeen polynarnaviruses containing 43: 753–78.
Arjona-Lopez, J. M. et al. (2018) ‘Novel, Diverse RNA Viruses from
multi-segmented genomes with their RdRp conserved domains
located in different segments. This finding suggests that multi- Mediterranean Isolates of the Phytopathogenic Fungus,
segmented polynarnaviruses are widely distributed in fungi, Rosellinia Necatrix: Insights into Evolutionary Biology of Fungal
and their genomes possibly contain up to seven segments. Viruses’, Environmental Microbiology, 20: 1464–83.
Based on the understanding of the lifestyle of multipartite vi- Arkhipova, K. et al. (2018) ‘Temporal Dynamics of Uncultured
ruses and multi-segmented viruses (Lowen 2018), the genome Viruses: A New Dimension in Viral Diversity’, The ISME Journal,
segmentation of polynarnaviruses may be advantageous for vi- 12: 199–211.
ral replication and recombination. We also found two narnavi- Attanayake, R. N. et al. (2013) ‘Sclerotinia sclerotiorum Populations
ruses, SsNV3 and SsNV4, with bi-segmented genomes. Based on Infecting Canola from China and the United States Are
Phyre2 results, the protein encoded by RNA 2 of SsNV3 showed Genetically and Phenotypically Distinct’, Phytopathology, 103:
the similarity to a -ssRNA virus, but I-TASSER results indicated 750–61.
the similarity to dsRNA viruses. We speculated that the differ- Ballaud, F. et al. (2015) ‘Dynamics of Viral Abundance and
ence between the two software results could be due to the poor Diversity in a Sphagnum-Dominated Peatland: Temporal
conservation of viral coat proteins, and the 3D structure of most Fluctuations Prevail over Habitat’, Frontiers in Microbiology, 6:
viral coat proteins has not been resolved, and there is no avail- 1494.
able adequate data related to the narnavirus coat proteins in Bankevich, A. et al. (2012) ‘SPAdes: A New Genome Assembly
the database. This may also be the reason why SsNV4 did not Algorithm and Its Applications to Single-Cell Sequencing’,
predict the information associated with the viral coat protein. Journal of Computational Biology: A Journal of Computational
We believed that more narnaviruses that could have coat pro- Molecular Cell Biology, 19: 455–77.
teins would be identified with the release of these two viral Bao, X., and Roossinck, M. J. (2013). Multiplexed Interactions:
sequences in the future. We speculated that these two narnavi- Viruses of Endophytic Fungi. Advances in Virus Research, 86: 37–58.
ruses may have obtained genes related to coat proteins from Bolger, A. M., Lohse, M., and Usadel, B. (2014) ‘Trimmomatic: A
other viruses or hosts, or that capsidless narnaviruses have lost Flexible Trimmer for Illumina Sequence Data’, Bioinformatics
their coat protein genes during the evolutionary process. These (Oxford, England), 30: 2114–20.
two viruses revealed an unanticipated evolutionary link be- Buchfink, B., Xie, C., and Huson, D. H. (2015) ‘Fast and Sensitive
tween encapsidated and capsidless RNA viruses. In summary, Protein Alignment Using DIAMOND’, Nature Methods, 12: 59–60.
these newly characterized mycoviruses with unique genome Capella-Gutierrez, S., Silla-Martinez, J. M., and Gabaldon, T.
features have a distant phylogenetic relationship from all (2009) ‘trimAl: A Tool for Automated Alignment Trimming in
known viruses and formed an independent evolutionary lineage Large-Scale Phylogenetic Analyses’, Bioinformatics, 25: 1972–3.
that supported the establishment of a new genus or family. Chen, C. et al. (2020) ‘TBtools: An Integrative Toolkit Developed
for Interactive Analyses of Big Biological Data’, Molecular Plant,
Acknowledgments 13: 1194–202.
Chiapello, M. et al. (2020) ‘Analysis of the Virome Associated to
We would like to thank Mr. Huang Huang in Huazhong Grapevine Downy Mildew Lesions Reveals New Mycovirus
Agricultural University and Mr. Zaiwu Zheng in the Plant Lineages’, Virus Evolution, 6: veaa058.
Protection Station of Wuxue City of Hubei Province for his Chiba, Y., Oiki S., Yaguchi T., Urayama S.-I. and Hagiwara D.
help in collecting samples, Dr. Kenichi Tsuda from (2020). ‘Discovery of divided RdRp sequences and a hitherto
Huazhong Agricultural University for constructive sugges- unknown genomic complexity in fungal viruses.’ Virus
tions, and Amanda Gutek for proof-reading the manuscript. Evolution, 7: veaa101.
This work was financially supported by the National Science Chow, C. E. T., and Fuhrman, J. A. (2012) ‘Seasonality and
Foundation of China (31722046), the National Key Research Monthly Dynamics of Marine Myovirus Communities’,
and Development Program of China (2017YFD0201100), and Environmental Microbiology, 14: 2171–83.
J. Jia et al. | 11

Deakin, G. et al. (2017) ‘Multiple Viral Infections in Agaricus bispo- Liu, L. et al. (2014) ‘Fungal Negative-Stranded RNA Virus That is
rus - Characterisation of 18 Unique RNA Viruses and 8 ORFans Related to Bornaviruses and Nyaviruses’, Proceedings of the
Identified by Deep Sequencing’, Scientific Reports, 7: 1–13. National Academy of Sciences of the United States of America, 111:
Dell’Anno, A., Corinaldesi, C., and Danovaro, R. (2015) ‘Virus 12205–10.
Decomposition Provides an Important Contribution to Benthic Lowen, A. C. (2018) ‘It’s in the Mix: Reassortment of Segmented
Deep-Sea Ecosystem Functioning’, Proceedings of the National Viral Genomes’, PLOS Pathogens, 14: e1007200.
Academy of Sciences, 112: E2014–E2019. Marquez, L. M. et al. (2007) ‘A Virus in a Fungus in a Plant:
Dolja, V. V., and Koonin, E. V. (2012) ‘Capsid-Less RNA Viruses’, Three-Way Symbiosis Required for Thermal Tolerance’,
in eLS. J. Wiley and L. Sons, Ltd: Chichester. Science, 315: 513–5.

Downloaded from https://academic.oup.com/ve/article/7/1/veab032/6206434 by Universidade Federal de Viçosa user on 25 March 2022


Farr, D. F., and Rossman, A. Y. (2020) ‘Fungal Databases’, U.S. Marzano, S.-Y. L. et al. (2016) ‘Identification of Diverse
National Fungus Collections, ARS, USDA. Mycoviruses through Metatranscriptomics Characterization of
Fu, L. et al. (2012) ‘CD-HIT: Accelerated for Clustering the the Viromes of Five Major Fungal Plant Pathogens’, Journal of
Next-Generation Sequencing Data’, Bioinformatics (Oxford, Virology, 90: 6846–63.
England), 28: 3150–2. Mu, F. et al. (2017) ‘Virome Characterization of a Collection of
Garcı́a-Pedrajas, M. et al. (2019) ‘Mycoviruses in Biological Sclerotinia sclerotiorum from Australia’, Frontiers in Microbiology,
Control: From Basic Research to Field Implementation’, 8: 2540.
Phytopathology, 109: 1828–39. Nakamura, T. et al. (2018) ‘Parallelization of MAFFT for
Ghabrial, S. A. et al. (2015) ‘50-plus Years of Fungal Viruses’, Large-Scale Multiple Sequence Alignments’, Bioinformatics
Virology, 479–480: 356–68. (Oxford, England), 34: 2490–2.
, and Suzuki, N. (2009) ‘Viruses of Plant Pathogenic Fungi’, Nerva, L. et al. (2019) ‘Mycoviruses Mediate Mycotoxin Regulation
Annual Review of Phytopathology, 47: 353–84. in Aspergillus Ochraceus’, Environmental Microbiology, 21: 1957–68.
Güemes, A. G. C. et al. (2016) ‘Viruses as Winners in the Game of Nerva, L. et al. (2019) ‘Isolation, Molecular Characterization and
Life’, Annual Review of Virology, 3: 197–214. Virome Analysis of Culturable Wood Fungal Endophytes in
Haas, B. J. et al. (2013) ‘De Novo Transcript Sequence Esca Symptomatic and Asymptomatic Grapevine Plants’,
Environmental Microbiology, 21: 2886–904.
Reconstruction from RNA-Seq Using the Trinity Platform for
Nguyen, L.-T. et al. (2015) ‘IQ-TREE: A Fast and Effective
Reference Generation and Analysis’, Nature Protocols, 8:
Stochastic Algorithm for Estimating Maximum-Likelihood
1494–512.
Phylogenies’, Molecular Biology and Evolution, 32: 268–74.
Helton, R. R. et al. (2012) ‘Interannual Dynamics of Viriobenthos
Nuss, D. L. (2005) ‘Hypovirulence: Mycoviruses at the
Abundance and Morphological Diversity in Chesapeake Bay
Fungal-Plant Interface’, Nature Reviews. Microbiology, 3: 632–42.
Sediments’, FEMS Microbiology Ecology, 79: 474–86.
Oerke, E. (2006) ‘Crop Losses to Pests’, The Journal of Agricultural
Ignacio-Espinoza, J. C., Ahlgren, N. A., and Fuhrman, J. A. (2020)
Science, 144: 31–43.
‘Long-Term Stability and Red Queen-like Strain Dynamics in
Ozkan, S., and Coutts, R. H. A. (2015) ‘Aspergillus fumigatus
Marine Viruses’, Nature Microbiology, 5: 265–71.
Mycovirus Causes Mild Hypervirulent Effect on Pathogenicity
Jiang, D. et al. (2013) ‘Viruses of the Plant Pathogenic Fungus
When Tested on Galleria mellonella’, Fungal Genetics and Biology:
Sclerotinia sclerotiorum’, Advances in Virus Research, 86: 215–48.
FG & B, 76: 20–6.
Kalyaanamoorthy, S. et al. (2017) ‘ModelFinder: Fast Model
Pearson, M. N. et al. (2009) ‘Mycoviruses of Filamentous Fungi
Selection for Accurate Phylogenetic Estimates’, Nature Methods,
and Their Relevance to Plant Pathology’, Molecular Plant
14: 587–9.
Pathology, 10: 115–28.
Kim, D., Langmead, B., and Salzberg, S. L. (2015) ‘HISAT: A Fast
Potgieter, A. C. et al. (2009) ‘Improved Strategies for
Spliced Aligner with Low Memory Requirements’, Nature
Sequence-Independent Amplification and Sequencing of Viral
Methods, 12: 357–60.
Double-Stranded RNA Genomes’, The Journal of General Virology,
Koonin, E. V., Senkevich, T. G., and Dolja, V. V. (2006)
90: 1423–32.
‘The Ancient Virus World and Evolution of Cells’, Biology Direct, Prussin, A. J. et al. (2019) ‘Seasonal Dynamics of DNA and RNA Viral
1: 29. Bioaerosol Communities in a Daycare Center’, Microbiome, 7: 53.
Kotta-Loizou, I., and Coutts, R. H. A. (2017) ‘Studies on the Qu, Z. et al. (2020) ‘Bio-Priming with a Hypovirulent
Virome of the Entomopathogenic Fungus Beauveria bassiana Phytopathogenic Fungus Enhances the Connection and
Reveal Novel dsRNA Elements and Mild Hypervirulence’, PLoS Strength of Microbial Interaction Network in Rapeseed’, Npj
Pathogens, 13: e1006183. Biofilms and Microbiomes, 6:
Krupovic, M., and Koonin, E. V. (2017) ‘Multiple Origins of Viral Rodriguez-Brito, B. et al. (2010) ‘Viral and Microbial Community
Capsid Proteins from Cellular Ancestors’, Proceedings of the Dynamics in Four Aquatic Environments’, The ISME Journal, 4:
National Academy of Sciences of the United States of America, 114: 739–51.
E2401–E2410. Sharma, P. et al. (2015) ‘Sclerotinia sclerotiorum (Lib) de Bary
Langmead, B., and Salzberg, S. L. (2012) ‘Fast Gapped-Read Causing Sclerotinia Rot in Oilseed Brassicas: A Review’, Journal
Alignment with Bowtie 2’, Nature Methods, 9: 357–9. of Oilseed Brassica, 6(Special Issue): 1–44.
Li, B., and Dewey, C. N. (2011) ‘RSEM: Accurate Transcript Sharon, A., and Shlezinger, N. (2013) ‘Fungi Infecting Plants and
Quantification from RNA-Seq Data with or without a Reference Animals: Killers, Non-Killers, and Cell Death’, PLoS Pathogens,
Genome’, BMC Bioinformatics, 12: 323. 9: e1003517.
Li, H., 1000 Genome Project Data Processing Subgroup. et al. Stöver, B. C., and Müller, K. F. (2010) ‘TreeGraph 2: Combining
(2009) ‘The Sequence Alignment/Map Format and SAMtools’, and Visualizing Evidence from Different Phylogenetic
Bioinformatics (Oxford, England), 25: 2078–9. Analyses’, BMC Bioinformatics, 11: 7.
Li, W. et al. (2015) ‘The EMBL-EBI Bioinformatics Web and Sutela, S. et al. (2020) ‘The Virome from a Collection of
Programmatic Tools Framework’, Nucleic Acids Research, 43: Endomycorrhizal Fungi Reveals New Viral Taxa with
W580–W584. Unprecedented Genome Organization’, Virus Evolution, 6: veaa076.
12 | Virus Evolution, 2021, Vol. 7, No. 1

Suttle, C. A. (2007) ‘Marine Viruses — Major Players in the Global Yu, X. et al. (2010) ‘A Geminivirus-Related DNA Mycovirus That
Ecosystem’, Nature Reviews. Microbiology, 5: 801–12. Confers Hypovirulence to a Plant Pathogenic Fungus’,
te Velthuis, A. J. (2014) ‘Common and Unique Features of Viral Proceedings of the National Academy of Sciences of the United States
RNA-Dependent Polymerases’, Cellular and Molecular Life of America, 107: 8387–92.
Sciences: CMLS, 71: 4403–20. et al. (2013) ‘Extracellular Transmission of a DNA
Thapa, V. et al. (2015) ‘Determinants of Taxonomic Composition Mycovirus and Its Use as a Natural Fungicide’, Proceedings of the
of Plant Viruses at the Nature Conservancy’s Tallgrass Prairie National Academy of Sciences of the United States of America, 110:
Preserve’, Virus Evolution, 1: vev007. 1452–7.
Whelan, S. P. J., Barr, J. N., and Wertz, G. W. (2004) ‘Transcription Zhang, H. et al. (2020) ‘A 2-kb Mycovirus Converts a Pathogenic

Downloaded from https://academic.oup.com/ve/article/7/1/veab032/6206434 by Universidade Federal de Viçosa user on 25 March 2022


and Replication of Nonsegmented Negative-Strand RNA Fungus into a Beneficial Endophyte for Brassica Protection and
Viruses’. Curr Top Microbiol Immunol, 283: 61–119. Yield Enhancement’, Molecular Plant, 13: 1420–33.
Wolf, Y. I. et al. (2018) ‘Origins And Evolution of the Global RNA Zhang, Y.-Z. et al. (2019) ‘Expanding the RNA Virosphere
Virome’, mBio, 9: e02329–02318. by Unbiased Metagenomics’, Annual Review of Virology, 6:
Xie, J., and Jiang, D. (2014) ‘New Insights into Mycoviruses and 119–39.
Exploration for the Biological Control of Crop Fungal Diseases’, , Shi, M., and Holmes, E. C. (2018) ‘Using Metagenomics to
Annual Review of Phytopathology, 52: 45–68. Characterize an Expanding Virosphere’, Cell, 172: 1168–72.

You might also like